probiotic bifidobacterium longum ncc3001 reduces depression …€¦ · t d accepted manuscript 1...

55
This is a repository copy of Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: a Pilot Study in Patients With Irritable Bowel Syndrome. White Rose Research Online URL for this paper: http://eprints.whiterose.ac.uk/116182/ Version: Accepted Version Article: Pinto-Sanchez, MI, Hall, GB, Ghajar, K et al. (19 more authors) (2017) Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: a Pilot Study in Patients With Irritable Bowel Syndrome. Gastroenterology, 153 (2). 448-459.e8. ISSN 0016-5085 https://doi.org/10.1053/j.gastro.2017.05.003 © 2017 by the AGA Institute. This manuscript version is made available under the CC-BY-NC-ND 4.0 license http://creativecommons.org/licenses/by-nc-nd/4.0/ [email protected] https://eprints.whiterose.ac.uk/ Reuse Items deposited in White Rose Research Online are protected by copyright, with all rights reserved unless indicated otherwise. They may be downloaded and/or printed for private study, or other acts as permitted by national copyright laws. The publisher or other rights holders may allow further reproduction and re-use of the full text version. This is indicated by the licence information on the White Rose Research Online record for the item. Takedown If you consider content in White Rose Research Online to be in breach of UK law, please notify us by emailing [email protected] including the URL of the record and the reason for the withdrawal request.

Upload: others

Post on 26-Jan-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

  • This is a repository copy of Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: a Pilot Study in Patients With Irritable Bowel Syndrome.

    White Rose Research Online URL for this paper:http://eprints.whiterose.ac.uk/116182/

    Version: Accepted Version

    Article:

    Pinto-Sanchez, MI, Hall, GB, Ghajar, K et al. (19 more authors) (2017) Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: aPilot Study in Patients With Irritable Bowel Syndrome. Gastroenterology, 153 (2). 448-459.e8. ISSN 0016-5085

    https://doi.org/10.1053/j.gastro.2017.05.003

    © 2017 by the AGA Institute. This manuscript version is made available under the CC-BY-NC-ND 4.0 license http://creativecommons.org/licenses/by-nc-nd/4.0/

    [email protected]://eprints.whiterose.ac.uk/

    Reuse

    Items deposited in White Rose Research Online are protected by copyright, with all rights reserved unless indicated otherwise. They may be downloaded and/or printed for private study, or other acts as permitted by national copyright laws. The publisher or other rights holders may allow further reproduction and re-use of the full text version. This is indicated by the licence information on the White Rose Research Online record for the item.

    Takedown

    If you consider content in White Rose Research Online to be in breach of UK law, please notify us by emailing [email protected] including the URL of the record and the reason for the withdrawal request.

    mailto:[email protected]://eprints.whiterose.ac.uk/

  • Accepted Manuscript

    Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and AltersBrain Activity: a Pilot Study in Patients With Irritable Bowel Syndrome

    Maria Ines Pinto-Sanchez, MD, Geoffrey B. Hall, PhD, Kathy Ghajar, BSc,Andrea Nardelli, MD, Carolina Bolino, MD, Jennifer T. Lau, BSc, Francois-PierreMartin, PhD, Ornella Cominetti, PhD, Christopher Welsh, BSc, Amber Rieder,BA, Jenna Traynor, BSc, Caitlin Gregory, MD, Giada De Palma, PhD, MarcPigrau, MD, Alexander C. Ford, MD, Joseph Macri, PhD, Bernard Berner, PhD,Gabriela Bergonzelli, PhD, Michael G. Surette, PhD, Stephen M. Collins, MD, PaulMoayyedi, MD, Premysl Bercik, MD.

    PII: S0016-5085(17)35557-9DOI: 10.1053/j.gastro.2017.05.003Reference: YGAST 61167

    To appear in: GastroenterologyAccepted Date: 2 May 2017

    Please cite this article as: Pinto-Sanchez MI, Hall GB, Ghajar K, Nardelli A, Bolino C, Lau JT, MartinF-P, Cominetti O, Welsh C, Rieder A, Traynor J, Gregory C, De Palma G, Pigrau M, Ford AC, MacriJ, Berner B, Bergonzelli G, Surette MG, Collins SM, Moayyedi P, Bercik P, Probiotic Bifidobacteriumlongum NCC3001 Reduces Depression Scores and Alters Brain Activity: a Pilot Study in Patients WithIrritable Bowel Syndrome, Gastroenterology (2017), doi: 10.1053/j.gastro.2017.05.003.

    This is a PDF file of an unedited manuscript that has been accepted for publication. As a service toour customers we are providing this early version of the manuscript. The manuscript will undergocopyediting, typesetting, and review of the resulting proof before it is published in its final form. Pleasenote that during the production process errors may be discovered which could affect the content, and alllegal disclaimers that apply to the journal pertain.

    http://dx.doi.org/10.1053/j.gastro.2017.05.003

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    1

    Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain

    Activity: a Pilot Study in Patients With Irritable Bowel Syndrome.

    Maria Ines Pinto-Sanchez1 MD, Geoffrey B Hall2 PhD, Kathy Ghajar2 BSc, Andrea Nardelli1 MD,

    Carolina Bolino1 MD, Jennifer T Lau1 BSc, Francois-Pierre Martin3 PhD, Ornella Cominetti3 PhD,

    Christopher Welsh1 BSc, Amber Rieder2 BA, Jenna Traynor2 BSc, Caitlin Gregory2 MD,

    Giada De Palma1 PhD, Marc Pigrau1 MD, Alexander C Ford4 MD, Joseph Macri5 PhD,

    Bernard Berner6 PhD, Gabriela Bergonzelli6 PhD, Michael G. Surette1 PhD,

    Stephen M Collins1 MD, Paul Moayyedi1 MD, Premysl Bercik1 MD.

    1Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster

    University, Hamilton, ON, Canada

    2Department of Psychology, Neuroscience & Behavior, McMaster University, Hamilton, ON,

    Canada.

    3Nestlé Institute of Health Sciences SA, Lausanne, Switzerland

    4Leeds Gastroenterology Institute, St. James's University Hospital, & Leeds Institute of Biomedical

    and Clinical Sciences, University of Leeds, Leeds, UK.

    5Department of Pathology, McMaster University, Hamilton, ON, Canada.

    6Nestlé Research Center, Nutrition Institute, Lausanne, Switzerland.

    Short Title: B. longum decreases depression in IBS patients

    Funding source: Nestlé SA, Switzerland,

    Corresponding author: Premysl Bercik, MD

    Department of Medicine, Farncombe Family Digestive Health Research Institute,

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    2

    McMaster University, Hamilton, ON, Canada

    e-mail: [email protected]

    CONFLICT OF INTEREST: MIPS, AN, GH, ACF, KG, AR, JT, CW, GDP, MP, JM, CG, CB,

    JL, MS and PM have no conflicts of interest concerning this paper. GB, and BB are employees of

    Nestec SA, Switzerland. MFP and OC are employees of Nestlé Institute of Health Sciences SA. PB

    and SMC received research support from Nestlé.

    DISCLOSURE: MIPS received a CIHR-CAG Health Professional Fellowship Award. PB is

    recipient of HHS Early Career Research Award and holder of Richard Hunt-AstraZeneca Chair in

    Gastroenterology.

    MANUSCRIPT CONTRIBUTION

    MIPS: acquisition, analysis and interpretation of data; statistical analysis, writing of the manuscript;

    GH: study concept and design; acquisition, analysis and interpretation of fMRI data; KG AR, JT,

    CG: fMRI data acquisition, analysis and interpretation; AN: clinical data acquisition, CB: study

    concept and design; CW: database development and data acquisition; JL, MS: microbiota analysis,

    critical review of manuscript; FPM, OC: NMR metabolomic analysis and data interpretation, BB:

    microbiota analysis, critical revision of the manuscript for important intellectual content, GB: study

    design, critical revision of the manuscript for important intellectual content: AF, SMC: critical

    review of the manuscript and important intellectual content; PM: Study design, statistical analysis,

    critical revision of the manuscript for important intellectual content; PB: Study design, critical

    revision of the manuscript and supervision of the study. All authors reviewed and approved the last

    version of the manuscript.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    3

    Acknowledgments:

    The authors thank Peter McLean and Mirna Del Valle for their invaluable support and study

    monitoring and oversight. We also thank Tiago Nunes for thoughtful comments on the manuscript,

    Mireille Goillard for neurotransmitter analysis, Francis Foata for the detection of BL and Margaret

    Fahnestock for advice on BDNF analysis and Laeticia Da Silva and Dr. Sebastiano Collino for

    their contribution in metabonomics data generation.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    4

    ABSTRACT

    Background & Aims: Probiotics can reduce symptoms of irritable bowel syndrome (IBS), but little

    is known about their effects on psychiatric comorbidities. We performed a prospective study to

    evaluate the effects of Bifidobacterium longum NCC3001 (BL) on anxiety and depression in

    patients with IBS.

    Methods: We performed a randomized, double-blind, placebo-controlled study of 44 adults with

    IBS and diarrhea or a mixed-stool pattern (based on Rome III criteria) and mild to moderate anxiety

    and/or depression (based on the Hospital Anxiety and Depression scale) at McMaster University in

    Canada, from March 2011 to May 2014. At the screening visit, clinical history and symptoms were

    assessed and blood samples were collected. Patients were then randomly assigned to groups and

    given daily BL (n=22) or placebo (n=22) for 6 weeks. At week 0, 6 and 10, we determined patients’

    levels of anxiety and depression, IBS symptoms, quality of life, and somatization using validated

    questionnaires. At week 0 and 6, stool, urine and blood samples were collected, and functional

    magnetic resonance imaging (fMRI) test was performed. We assessed brain activation patterns,

    fecal microbiota, urine metabolome profiles, serum markers of inflammation, neurotransmitters and

    neurotrophin levels.

    Results: At week 6, 14/22 patients in the BL group had reduction in depression scores of 2 points or

    more on the Hospital Anxiety and Depression scale, vs 7/22 patients in the placebo group (P=.04).

    BL had no significant effect on anxiety or IBS symptoms. Patients in the BL group had a mean

    increase in quality of life score compared with the placebo group. The fMRI analysis showed that

    BL reduced responses to negative emotional stimuli in multiple brain areas, including amygdala and

    fronto–limbic regions, compared with placebo. The groups had similar fecal microbiota profiles,

    serum markers of inflammation, and levels of neurotrophins and neurotransmitters, but the BL

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    5

    group had reduced urine levels of methylamines and aromatic amino acids metabolites. At week 10,

    depression scores were reduced in patients given BL vs placebo.

    Conclusion: In a placebo-controlled trial, we found that the probiotic BL reduces depression but not

    anxiety scores and increases quality of life in patients with IBS. These improvements were

    associated with changes in brain activation patterns that indicate that this probiotic reduces limbic

    reactivity. ClinicalTrials.gov no. NCT01276626.

    Key words: IBS, anxiety, depression, fMRI

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    6

    BACKGROUND

    Irritable bowel syndrome (IBS), characterized by abdominal pain and altered bowel habits, affects

    11% of the world-wide population1, has a significant socioeconomic impact2 and its current

    treatments have limited efficacy1. Its pathophysiology is incompletely understood but is considered

    to be a disorder of gut-brain interaction3 and is frequently accompanied by psychiatric disorders1,4.

    Accumulating evidence suggests that commensal bacteria play a role in IBS, as multiple studies

    have demonstrated an abnormal composition or metabolic activity of gut microbiota in patients with

    IBS5. Dysbiosis, triggered by acute bacterial gastroenteritis, antibiotics or dietary factors, which are

    known to affect the composition of microbiota, may drive not only the gastrointestinal component

    of IBS but also contribute to its psychiatric comorbidity6. Furthermore, specific probiotic bacteria

    have been shown to improve gastrointestinal symptoms in IBS7.

    We have previously demonstrated that administration of B. longum NCC3001 subspecies longum

    strain (BL) normalized anxiety-like behavior and hippocampal Brain Derived Neurotrophic Factor

    (BDNF) levels in mice with low-grade gut inflammation, through vagal dependent pathways8,9.

    Based on these results, we hypothesized that BL will improve psychiatric comorbidity in patients

    with chronic bowel disorders and thus we performed a pilot study in IBS patients. As anxiety and

    depression are rather difficult to distinguish in animal models, they frequently co-exist in patients

    and altered central BDNF levels were reported in both conditions, we chose as our primary

    objective to evaluate the impact of BL on co-morbid anxiety and depression. The secondary

    objectives were then to assess the effect of BL on IBS symptoms and quality of life, and to explore

    changes in brain activation patterns, circulating inflammatory markers, neurotransmitters,

    neurotrophins, gut microbiota profile and urine metabolites as a measure of host-microbial

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    7

    metabolic interactions. Considering the large heterogeneity of IBS we decided to restrict our study

    to IBS patients with diarrhea or mixed stool pattern, as they apper to share similar sensory neuro-

    imune interaction and are more likely to present with low-grade gut inflammation and similar

    microbiota compared with IBS patients with constipation10,5.

    Although several clinical studies investigated effects of probiotic bacteria on behavior and brain

    function11-12, mostly in healthy individuals, our study is the first one to show that probiotics can

    improve depression scores as well as alter brain activity patterns in IBS patients with comorbid

    depression and anxiety.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    8

    METHODS

    Study oversight

    We conducted a randomized, double-blind, placebo-controlled, single center pilot study from

    March 2011 to May 2014. The study was approved by the Hamilton Health Sciences and St.

    Joseph’s Health Care Research Ethics Boards, all participants signed the informed consent. The

    study was registered in clinicaltrials.gov under NCT01276626. All authors had access to the study

    data and reviewed and approved the final manuscript.

    Participants

    We recruited adult patients with a diagnosis of IBS with diarrhea or mixed-stool pattern (Rome III

    criteria)13, and mild to moderate anxiety and/or depression scores based on the Hospital Anxiety

    and Depression (HAD) scale14 (HAD-A or HAD-D score 8-14). Patients with a history of organic

    diseases, immune deficiency, major abdominal surgery, a psychiatric condition other than anxiety

    or depression, use of immunosuppressants, glucocorticosteroids, opioids, antidepressants or

    anxiolytics in regular doses, alcohol or illicit drug consumption, were excluded. Loperamide and

    laxatives were allowed as rescue medications. Other probiotics in any form were forbidden during

    the 1-month run-in period and the trial. Antibiotics were forbidden during the 3 months prior to the

    run-in period and the trial.

    Design of the study

    The study involved four hospital visits (Supplementary figure 1). At the screening visit ( -4 weeks),

    clinical history and symptoms were assessed and physical exam and complete bloodwork

    performed. At the baseline visit (week 0), the inclusion and exclusion criteria and symptoms were

    re-assessed, stool, urine and blood samples were collected, and an fMRI study performed.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    9

    The patients were then randomised to receive 42 sachets of either spray dried BL (1.0E+10 CFU

    /1gram powder with maltodextrin) or placebo containing 1 gram of maltodextrin. Treatment

    products were indistinguishable in terms of package, color, taste and consistency. Patients were

    instructed to dissolve the content of the sachet in 100-200 ml of lactose-free milk, soy milk or rice

    milk, preheated to 20° Celsius. Patients were asked not to change their eating habits or fibre intake.

    Participants recorded the treatment intake, the empty sachets were used to assess the compliance at

    the next visit (week 6), where their symptoms were assessed, blood, urine and stool samples

    collected and fMRI test performed. Finally, patients’ symptoms were re-assessed at a follow-up

    visit (week 10).

    In addition to the regular hospital visits, HAD scores were also assessed at 3 weeks of treatment

    following request of Health Canada. HAD questionnaires were provided to patients at Visit 1 and

    then mailed or e-mailed to the investigators.

    Study endpoints

    The primary endpoint was a reduction in anxiety and/or depression scores of ≥2 points on HAD

    scale13 at 6 weeks. This was based on the previously established mean clinically important

    difference for the anxiety and depression score on the HAD scale of 1.3 and 1.4, respectively14.

    Secondary endpoints included improvement in anxiety and depression scores (HAD, continuous

    data), anxiety (State-Trait Anxiety Inventory, STAI), IBS global adequate relief, IBS symptoms,

    somatization, quality of life, changes in brain activation patterns (functional Magnetic Resonance

    Imaging, fMRI), serum inflammatory markers, neurotransmitters and BDNF, and urine

    metabonomic and stool microbiota profiles.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    10

    Randomization

    The randomization sequence was performed using a computer program (Proc Plan, SAS, V. 9.1). A

    block randomization was stratified by gender and IBS status (diarrhea or mixed stool pattern). The

    codes were kept in sealed opaque envelopes allocated to patients according to strata. Each pack was

    assigned a number according to the randomization sequence. On recruitment, the patients were

    assigned into one of four strata and given the next consecutive randomization number available for

    that stratum. Treatment allocation was concealed from participants and study staff.

    Treatment products indistinguishable in terms of package, color, taste and consistency, were

    identified with two non-speaking codes per arm. Their identity was blind to subjects, investigators

    and support staff, known only by the manufacturer, Nestlé Product Technology Centre Konolfingen

    Switzerland.

    Study Measurements

    Anxiety and depression were assessed by the HAD score. As an additional measure of anxiety we

    used the STAI15, which assesses both state and trait anxiety. IBS symptoms and signs were assessed

    by the Birmingham IBS score16 and Bristol stool scale17. To evaluate an overall improvement of

    IBS symptoms, patients were asked a validated question: “Over the past week have you had

    adequate relief of your IBS symptoms?” with a dichotomous option for responses18. Health-related

    quality of life (QoL) was measured by the SF-3619 and somatization by the PHQ-15

    questionnaires20.

    Brain activity was assessed by functional magnetic resonance imaging (fMRI) using General

    Electric 3-Tesla Discovery MR 750, whole body short bore scanner with 32 parallel receiver

    channels (General Electric, Milwaukee, WI). The 1-hour protocol included a seven minute T1

    weighted structural scan, followed by four repetitions of a fearful face backward masking

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    11

    paradigm21 (Supplementary figure 2) during four fMRI Blood Oxygen Level Dependent scans22

    (BOLD EPI; TR/TE=2800/35 ms, flip angle=90º, 3 mm thick slices, no gap, field of view=24 cm,

    matrix=64x64). Pre-processing of MRI data was completed using Brain Voyager QX Version 2.8.2,

    32-bit (Brain Innovation, Maastricht, Netherlands). Anatomic and functional data were inspected

    and scans with artefacts or fMRI scans with movement greater than 5 mm in any of 6 planes were

    excluded from analysis. Anatomical scans were transformed into standard sagittal orientation, and

    underwent spatial normalization into standard Talaraich space. Slice scan time correction and 3D

    motion correction were carried out on the fMRI data and spatial smoothing applied using a

    Gaussian filter (FWHM=6 mm). The amygdala was selected as region of interest (ROI), initially

    derived from the WFUPick Atlas and refined according to anatomic landmarks on the full group

    average transformed T1 image.

    Blood and urine samples were collected after an overnight fast. After processing, the samples were

    stored at -80 C until assessment. Samples for BDNF were collected using the PAXgene Blood RNA

    (PreAnalytiX, Qiagen BD, Toronto, Canada). Serum cytokines and CRP levels were assessed by

    Human ProInflammatory 7-Plex Ultra-Sensitive Kit (MSD, Gaithersburg, MD) and CRP Abbott

    Architect kit (Abbott Laboratories, IL), respectively. BDNF protein level was assessed by Human

    BDNF DuoSet ELISA kit (R&D Systems, Minneapolis, MN). Plasma neurotransmitters were

    quantified using following kits: 5-HT: IBL, Hamburg, Germany; Substance P: Abcam, Cambridge,

    UK; CGRP: Cloud-Clone Corp, Houston, TX.

    Urine metabolites were assessed by 1H NMR profiling using a Bruker Avance II 600 MHz

    spectrometer equipped with a 1.7 mm probe at 300 K (Bruker Biospin, Rheinstetten, Germany),

    using a standard pulse sequence with water suppression, and processed using TOPSPIN (version

    2.1, Bruker, Germany) software package. The metabolite identification was achieved using in house

    database and 2D 1H NMR spectroscopy experiments. Chemometric analysis was performed using

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    12

    the software package SIMCA-P+ (version 14.0, Umetrics AB, Umeå, Sweden) and in-house

    developed MATLAB routines. Orthogonal Projection to Latent Structures (OPLS) and OPLS

    discriminant analysis (OPLS-DA) were employed for exploring the variance in the metabonomics

    data that may explain statistical differences between groups of samples. The classification accuracy

    of the OPLS-DA was established from the predicted samples in the 7-fold cross-validation cycle.

    To highlight the weight of individual variables in the model, Variable Importance in Projection

    (VIP) was used, with a value above 1 used as a threshold by convention. For additional details, see

    Supplementary Methods.

    Microbiota analysis was performed using Illumina sequencing of the V3 region of 16S rRNA gene

    as described previously23, for details see Supplementary Methods. Bacterial strain-specific PCR24

    was used on fecal DNA extracts to detect the presence of BL at the end of the treatment period.

    Statistical analysis

    Statistical analyses were performed using IBM-SPSS (IBM-SPSS Statistics v20, Chicago, IL). We

    performed post-hoc power calculations based on our previous animal data, which showed strong

    therapeutic potential of this probiotic8-9. We estimated that a sample size of 19 in each group would

    have 80% power using a two-group χ2 test with 0.05 two-sided significance level assuming 30%

    have an improvement in depression and/or anxiety in the placebo group and 75% in the B. longum

    group.

    Data from all randomized subjects were analyzed according to intention to treat (ITT) principles for

    the primary outcome. To deal with missing data, we used the extreme case analysis assuming that

    all missing subjects had no improvement in symptoms. Per protocol evaluation (PP) excluded data

    from subjects who did not complete the trial due to consumption of proscribed medication or non-

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    13

    compliance with the study protocol, and was used for the primary and secondary outcomes. For

    testing the effects on the two primary endpoints, Pearson Chi-Squared and Mann-Whitney U test

    were used as appropriate. In addition, the HAD scores were analyzed at baseline, week 3, 6 and 10

    post-treatment using ANOVA repeated measures. ANCOVA was used to adjust for baselines

    differences in HAD depression scores. A two-sided test was used and p

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    14

    Nestec SA was not involved in collection, analysis or interpretation of the clinical data. The

    corresponding author was in charge of collection and analysis of data and had final responsibility

    for the decision to submit for publication.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    15

    RESULTS

    Study patients

    Sixty patients were enrolled in the study (Figure 1). Sixteen patients failed the screening due to: 1)

    very mild IBS symptoms (n=2) or low HAD score (n=1); 2) use of antibiotics (n=2); 3)

    unwillingness to follow the protocol (n=7), or 5) loss to follow-up (n=4). Thus, 44 patients were

    randomized (22 in each study arm), from whom 38 completed the study (BL=18, placebo=20).

    During the treatment, six patients dropped out due to use of antibiotics (n=4), or antidepressants

    (n=2). No differences were observed between the groups in baseline characteristics (Table 1),

    except for higher HAD-D scores in the BL group (Table 2).

    Primary outcome: Improvement in depression and/or anxiety scores

    At 6 weeks, 14 out of 22 (64%) patients in the BL group had decreased depression scores (HAD-D

    ≥2 points) compared with 7 out of 22 (32%) patients given placebo (relative risk (RR) 1.98; 95%

    confidence interval (CI) 1.16-3.38; p=0.04) in the ITT population (Table 3A). This response was

    more prominent in the PP analysis with 78% in the BL group compared with 35% of the placebo

    group having lower depression scores (RR 2.4; 95% CI 1.26-4.58; p=0.016). The improvement in

    HAD-D scores was sustained in both the ITT (RR 2.05; 95% CI 1.07-3.93; p=0.04) and the PP

    analysis (RR 2.14; 95% CI 1.11-4.12; p=0.04) at 10-week follow-up. The results at 6 weeks were

    similar when the analysis was performed in only the subgroup of patients with baseline scores

    indicative of depression (HAD-D ≥8), (RR 3.75; 95% CI 0.6-22.1; p=0.047). No significant

    differences in number of patients with decreased anxiety (HAD-A ≥2 points) were found between

    the groups at 6 or 10-week follow-up.

    A sensitivity analysis performed to explore the relationship between depression scores and

    gastrointestinal symptoms showed that the beneficial effect of BL on depression scores at 6 and 10

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    16

    weeks was more likely to occur in those patients who reported adequate relief of IBS symptoms

    (Table 3B).

    Secondary outcomes

    HAD scores

    After treatment, there were no significant differences on HAD-D scores when assessed as a

    continuous outcome between the two groups (Table 2), but when adjusting for baseline differences

    a greater improvement was found in the BL group (ANCOVA p=0.049). Anxiety scores assessed as

    a continuous outcome were similar between the two groups (Table 2).

    IBS symptoms

    There was no difference in adequate relief of IBS symptoms in the ITT analysis at 6 weeks (RR 1.6,

    95% CI 0.86-2.91; p=0.22) but the PP analysis showed a statistically significant benefit of BL over

    placebo (RR 2.03 95% CI 1.13-3.65; p=0.02). This beneficial effect was not, however, maintained

    at 10-week follow-up (RR 0.70, 95% CI 0.24-2.09; p=0.52). No significant differences were

    observed in the overall Birmingham score or sub scores for constipation, diarrhea or pain at 6 or 10

    weeks (Table 2).

    State and trait anxiety and somatization

    No differences in STAI scores were observed between groups at 6-week or 10-week follow-up

    (Table 2). Somatization scores were also similar between groups at 6-week or 10-week follow up

    (Table 2).

    Quality of life

    QoL improved in the physical subdomain in the BL group compared with placebo (p=0.03; 95% CI

    0.01-0.90) (Table 2), with amelioration in general physical health (physical functioning; p=0.04;

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    17

    95% CI 0.43-0.51) and problems with work or other daily activities (role physical; p=0.01; 95% CI

    0.009-0.013).

    Brain activation patterns by fMRI

    Brain activation patterns differed in response to fearful versus happy faces in all subjects studied

    (Supplementary Figure 3, Supplementary Table 1). Before treatment, there was no difference in

    response to fear stimuli versus fixation between placebo and the BL group, except for greater

    engagement of the visual association and parietal cortices in the latter group (Figure 2A-B).

    However, after treatment, compared with placebo, the BL group showed reduced engagement of the

    amygdala, frontal and temporal cortices, as well as heightened engagement of occipital regions in

    response to the fear stimuli (compared with fixation) (Figure 2B-C, Table 4). Overall, the change in

    engagement of the amygdala correlated with the change in depression scores (r=0.52, p=0.004;

    Figure 2E). Within the BL group, reduced engagement of the amygdala correlated with decreased

    depression scores (r=0.58, p=0.03), but this was not observed in the placebo group (r=0.20,

    p=0.46). In the BL group, reduced engagement of the amygdala was more likely to occur in patients

    with adequate relief of IBS symptoms than in those without it (RR 3.07 95% CI 0.89-10.59; p=0.03),

    but this was not observed in the control group (RR 1.5 95% CI 1.00-2.23; p=0.51; Figure 2E).

    There was no difference when comparing response to fearful versus neutral faces.

    Inflammatory markers, neurotransmitters and BDNF levels

    No differences in serum inflammatory markers (CRP, TNF-α, IFN-γ, IL-1β, IL-6, IL-8, IL-10, IL12

    and IL-10/12 ratio) or neurotransmitters (5-HT, substance P and CGRP) were found between the

    groups at 6 weeks (Supplementary table 2). Similarly, no changes in blood BDNF mRNA or

    BDNF protein levels were identified.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    18

    Intestinal microbiota composition

    There were no major differences in taxa, compositional distance or alpha diversity (Shannon, Chao1

    and Observed Species indices) before or after the treatment between the two groups. Microbiota

    profiles were also similar when assessed by Bray-Curtis Principle Coordinate analysis or Bray-

    Curtis Distance comparisons (Supplementary Figure 4). BL was detected at the end of treatment in

    15 out of 18 (80%) patients of the probiotic group.

    Urine metabonomics

    OPLS discriminant analysis was applied using one predictive and two orthogonal components to

    model urine metabolic differences between the two groups (Supplementary Figure 5). The model

    was statistically robust only for post-treatment analysis (R2X=0.17, R2Y=0.84, Q2Y=0.20, where

    R2X: explained variance in the metabonomics data (urine metabolites), R2Y: explained group

    variance (placebo and probiotic) and Q2Y: robustness of the model). Before treatment, there was

    no difference between the two groups (Q2Y

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    19

    DISCUSSION

    In this randomized, placebo controlled study we found that 6-week administration of

    Bifidobacterium longum NCC3001 (BL) decreased depression but not anxiety scores, which was

    our primary outcome, and decreased responses to fearful stimuli in multiple brain areas involved in

    the processing of emotions, including the amygdala and fronto–limbic regions. Patients given BL

    also reported improvement in overall symptoms of IBS and physical domain of QoL. Despite being

    a pilot study with limited subject numbers, this is the first trial to show that a specific probiotic

    improves depression scores in IBS patients and induces pronounced changes in brain activity in

    regions that have been previously implicated in depression, and that are influenced by anti-

    depressant therapy27,28. Indeed, both depression and anxiety disorders have been associated with

    amygdala hyperactivity and converging evidence suggests that one mechanism by which Selective

    Serotonin Re-uptake Inhibitors (SSRIs) exert their beneficial action is by downregulating its

    activity27,28. Our study also validates the use of some murine models8,9 to screen probiotics for their

    potential therapeutic benefit in humans, as BL was shown previously to improve anxiety-like

    behavior and brain chemistry in mice.

    There is growing interest in the role of the intestinal microbiota in health and disease. Gut bacteria

    not only instruct and shape the host immune system, and impact its metabolism, but also affect

    function in the gut and central nervous system29,30. Animal studies have demonstrated that changes

    in microbiota composition31 or administration of specific probiotics, including bifidobacteria, can

    alter behavior and brain chemistry of the host8,9,32. Multiple mechanisms are likely involved in this

    microbiota-brain communication, including immune, neural and metabolic pathways30. We have

    previously extensively investigated the beneficial effect of BL on behavior in murine models and

    shown that this probiotic normalizes inflammation-induced anxiety-like behavior and hippocampal

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    20

    BDNF levels8,9. The beneficial effect on behavior was mediated through vagal signaling, possibly

    through release of neuroactive metabolites acting on enteric neurons9.

    Despite accumulating evidence of the influence of microbiota on behavior in animal models, data

    from humans are rather limited30. Two studies in healthy volunteers reported no major effects of

    probiotic on anxiety and depression scores33,34. A very recent pilot study in patients with major

    depression showed that depression improved in both the placebo and probiotic groups, although the

    improvement appeared to be greater in the latter35. The only study, which directly linked the gut

    microbiota to brain function, assessed effects of mixture of probiotic bacteria in healthy females.

    Although no change in behavior in that trial was observed, fMRI showed decreased BOLD activity

    in the limbic and sensory brain regions in response to facial expressions, which stimulate emotional

    responses12. We used a similar emotive challenge that is known to induce activation in several brain

    regions, including the amygdala, where the fear response is particularly salient36, and which was

    thus chosen as a predetermined ROI. We found that BL administration decreased activation of the

    amygdala and the fronto–limbic complex compared with placebo, which was paralleled by

    decreased depression scores. The amygdala is not only involved in regulation of fear and anxiety,

    but also in activation of the hypothalamic-pituitary-adrenal axis (HPA)37 and modulation of the

    visceral sensitivity38. In our study, pain scores appeared to improve more in the BL group compared

    to the placebo group, although the difference was not statistically significant. However, the BL

    group was more likely to report the adequate relief of IBS symptoms and this was associated with

    decreased amygdala activation. Thus changes in pain perception could explain the overall

    improvement of IBS symptom in the BL group but this should be further investigated in future,

    better powered trials.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    21

    Multiple studies have demonstrated beneficial effects of different probiotics in patients with IBS,

    and a recent meta-analysis7 has shown that bifidobacteria are more likely to improve

    gastrointestinal symptoms than lactobacilli. Underlying mechanisms may involve improvement of

    low-grade gut inflammation present in a subset of patients with IBS39. Inflammation has also been

    linked to psychiatric disorders as pro-inflammatory cytokines can affect mood by a number of

    mechanisms, including activation of the HPA axis and alteration of the metabolism of

    neurotransmitters40. We found no differences in serum CRP or cytokines levels between placebo

    and patients given BL, indicating that the beneficial effect of this probiotic is not mediated by

    improvement in inflammation, in agreement with the previous study in mice8.

    Our murine study demonstrated that hippocampal BDNF was upregulated by BL8. Neurotrophin

    BDNF influences survival and differentiation of neurons, formation of functional synapses and

    neuroplasticity41 and is decreased in major depressive disorder42. We found no differences in serum

    BDNF or neurotransmitter levels, including serotonin, in patients given BL compared with patients

    in the placebo group but this does not rule out the possibility that BDNF or neutransmitters levels

    in specific brain regions were modulated by the probiotic.

    As changes in gut microbiota composition have been linked to altered behavior and brain

    chemistry31, we assessed fecal bacterial profiles before and after the treatment. Analysis of fecal

    16S rRNA gene sequencing suggests that the beneficial effect of BL is independent of major

    alterations in microbioal composition. However, BL altered the urinary metabolic profile suggestive

    of downregulated bacterial degradation of methylamines and aromatic amino acids43, including a

    decrease in host-bacterial co-metabolite 4-cresol sulfate, a by-product of tyrosine fermentation44.

    Interestingly, 4-cresol sulfate is known to inhibit dopamine β-hydroxylase45, which is a constituent

    of the catecholamine biosynthetic pathway involved in mood regulation46. This enzyme converting

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    22

    dopamine into noradrenaline is expressed in the central and peripheral nervous system as well as in

    chromaffin cells of the adrenal medulla47, and its decreased activity has been associated with

    depression46. In our study, levels of 4-cresol sulfate correlated with improved depression scores in

    BL group suggesting that dopamine/noradrenaline pathway may play an important role in the effect

    of this probiotic. In future studies, a targeted metabolomic approach should provide further insight

    into the impact of BL on the bacterial metabolism of aromatic amino acids and the subsequent

    modulation of the host’s catecholamine production.

    Although this is the first randomized trial to show that probiotics decrease depression scores in IBS

    patients there are limitations that are important to emphasize. We used the HAD scale to identify

    patients with psychiatric comorbidity, which is well validated and widely used in studies

    investigating IBS as a simple tool to screen for comorbid anxiety or depression3. Its psychometric

    properties may be inferior to clinician-administered rating scales for depression and anxiety, but its

    main advantage is that it does not measure somatic symptoms13,48, a strong confounding factor in

    any study of IBS patients. Indeed, there is no superior validated questionnaire that can be used in

    patients with a concurrent medical disorder. However, a confirmatory trial using additional specific

    psychometric tools in a larger cohort of patient is needed to substantiate our current results. A

    weakness of our study was the difference in the baseline depression scores between the two groups

    with lower values in the placebo group. The improvement in depression in the probiotic group

    could relate to regression to the mean or reflect a floor effect. We believe this is unlikely, however,

    as a statistically significant result in favor of BL remained when adjusting for baseline differences

    using, and when performing analysis only in the subgroup of patients with baseline scores

    indicative of depression (HAD-D ≥8). Also, there was a linear decrease in the depression subscale

    of the HAD score in the BL group that did not asymptote with the placebo HAD score

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    23

    (Supplementary Figure 6). Furthermore, the observed changes in brain activation patterns in the

    areas involved in mood regulation in the BL group support the notion that this probiotic has anti-

    depressive properties. We have not shown any effect of BL on individual gut symptoms but our

    study was not adequately powered to detect these changes. Thus, a larger, appropriately powered

    trial with patients with IBS and comorbid depression is needed to verify our data on psychiatric and

    gut symptoms before BL can be recommended in clinical practice.

    In conclusion, B. longum NC3001 has longstanding beneficial effects on mild to moderate comorbid

    depression and it temporarily improves overall symptoms of IBS and QoL. This is associated with

    changes in activity of multiple brain areas involved in emotional processing, but no improvement in

    individual gut symptoms, suggesting that central effects of this probiotic underlies its therapeutic

    effect, possibly through modulation of host catecholamine production.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    24

    REFERENCES

    1- Lacy BE, Mearin F, Chang L, et al. Bowel disorders. Gastroenterology 2016; 150(6): 1393–

    1407.

    2- Canavan C, West J, Card T. Review article: the economic impact of the irritable bowel

    syndrome. Aliment Pharmacol Ther 2014; 40(9): 1023-34.

    3- Drossman DA, Hasler WL. Rome IV—Functional GI disorders: Disorders of gut-brain

    interaction. Gastroenterology 2016; 150(6): 1257–1261.

    4- Palsson OS, Drossman DA. Psychiatric and psychological dysfunction in irritable bowel

    syndrome and the role of psychological treatments. Gastroenterol Clin North Am 2005; 34(2): 281-

    303.

    5- Simrén M, Barbara G, Flint HJ, et al. Intestinal microbiota in functional bowel disorders: a Rome

    foundation report. Gut 2013; 62: 159-76.

    6- Barbara G, Feinle-Bisset C, Ghoshal EC, et al. The intestinal microenvironment and functional

    gastrointestinal disorders. Gastroenterology 2016; 150(6): 1305–1318.

    7- Ford AC, Quigley EMM, Lacy BE, et al. Efficacy of prebiotics, probiotics, and synbiotics in

    Irritable Bowel Syndrome and chronic idiopathic constipation: Systematic review and meta-

    analysis. Am J Gastroenterol 2014; 109:1547–61.

    8- Bercik P, Verdu EF, Foster JA, et al. Chronic gastrointestinal inflammation induces anxiety-like

    behavior and alters central nervous system biochemistry in mice. Gastroenterology 2010; 139(6):

    2102-12.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    25

    9- Bercik P, Park AJ, Sinclair D, et al. The anxiolytic effect of Bifidobacterium longum NCC3001

    involves vagal pathways for gut-brain communication. Neurogastroenterol Motil 2011; 23(12):

    1132-9.

    10- Hughes PA, Harrington AM, Castro J, et al. Sensory neuro-immune interactions differ between

    irritable bowel syndrome subtypes. Gut 2013; 62(10): 1456-65.

    11- McKean J, Naug H, Nikbakht E, et al. Probiotics and Subclinical Psychological Symptoms in

    Healthy Participants: A Systematic Review and Meta-Analysis. J Altern Complement Med 2016

    Nov 14.

    12-Tillisch K, Labus J, Kilpatrick, et al. Consumption of fermented milk product with probiotic

    modulates brain activity. Gastroenterology 2013; 144(7): 1394-401.

    13- Longstreth GF, Thompson WG, Chey WD, et al. Functional bowel disorders. Gastroenterology

    2006; 130(5): 1480-91.

    14- Snaith RP, Zigmond AS. The Hospital anxiety and depression scale with the Irritability

    depression – anxiety scale and the Leeds situational anxiety scale manual. Published by GL

    assessment Ltd. 1994.

    15- Puhan M, Frey M, Büchi S, et al. The minimal important difference of the hospital anxiety and

    depression scale in patients with chronic obstructive pulmonary disease. Health Qual Life Outcomes

    2008; 6: 46.

    16- Gaudry E, Spielberger CD, Vagg P. Validation of state-trait distinction in anxiety distinction.

    Multivariate Behav Res 1975;10:331–41.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    26

    16- Roalfe AK, Roberts LM, Wilson S. Evaluation of the Birmingham IBS symptom questionnaire.

    BMC Gastroenterol 2008 Jul 23; 8:30.

    17- O'Donnell LJ, Virjee J, Heaton KW. Detection of pseudodiarrhoea by simple clinical

    assessment of intestinal transit rate. mBMJ 1990; 300(6722): 439-40.

    18- Passos MC, Lembo AJ, Conboy LA, et al. Adequate relief in a treatment trial with IBS patients:

    a prospective assessment. Am J Gastroenterol 2009; 104(4): 912–9.

    19- McHorney CA, Ware JE Jr, Raczek AE. The MOS 36-Item Short-Form Health Survey (SF-36):

    II. Psychometric and clinical tests of validity in measuring physical and mental health constructs.

    Med Care 1993;31(3):247-63.

    20- Kroenke K, Spitzer RL, Williams JB. The PHQ-15: validity of a new measure for evaluating the

    severity of somatic symptoms. Psychosom Med 2002; 64(2): 258-66.

    21- Hall GB, Doyle KA, Goldberg J, et al. Amygdala engagement in response to subthreshold

    presentations of anxious face stimuli in adults with Autism Spectrum Disorders: preliminary

    insights. PloS One 2010; 5(5): e10804.

    22- He X, Yablonskiy DA. Quantitative BOLD: mapping of human cerebral deoxygenated blood

    volume and oxygen extraction fraction: default state. Magn Reson Med 2007; 57:115–26.

    23- De Palma G, Blennerhassett P, Lu J, et al. Microbiota and host determinants of behavioral

    phenotype in maternally separated mice. Nat Commun 2015; 6: 7735.

    24- Rougé C, Goldenberg O, Ferraris L, et al. Investigation of the intestinal microbiota in preterm

    infants using different methods. Anaerobe 2010; 16(4): 362-70.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    27

    25- Genovese CR, Lazar NA, Nichols T. Thresholding of statistical maps in functional

    neuroimaging using the false discovery rate. Neuroimage 2002;15(4): 870-8.

    26- Maldjian JA, Laurienti PJ, Kraft RA, Burdette JH. An automated method for neuroanatomic

    and cytoarchitectonic atlas-based interrogation of fmri data sets. NeuroImage 2003; 19, 1233–

    1239.

    27- Harmer CJ, Mackay CE, Reid CB, et al. Antidepressant drug treatment modifies the neural

    processing of nonconscious threat cues. Biol Psychiatry 2006; 59(9): 816-20.

    28- Fu CH, Williams SC, Cleare AJ, et al. Attenuation of the neural response to sad faces in major

    depression by antidepressant treatment: a prospective, event-related functional magnetic resonance

    imaging study. Arch Gen Psychiatry 2004; 61: 877–89.

    29- Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune

    system. Science 2012; 336 (6086): 1268-73.

    30- Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain.

    Nat Rev Microbiol 2012; 10(11): 735-42.

    31- Bercik P, Denou E, Collins J, et al. The intestinal microbiota affect central levels of brain-

    derived neurotropic factor and behavior in mice. Gastroenterology 2011; 141(2): 599–60.

    32- Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus strain regulates emotional

    behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci

    U S A 2011; 108(38): 16050-5.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    28

    33- Messaoudi M, Violle N, Bisson JF, et al. Beneficial psychological effects of a probiotic

    formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human

    volunteers. Gut Microbes 2011;2(4):256-61

    34- Benton D, Williams C, Brown A. Impact of consuming a milk drink containing a probiotic on

    mood and cognition. Eur J Clin Nutr 2007; 61(3):355-61.

    35- Akkasheh G, Kashani-Poor Z, Tajabadi-Ebrahimi M, et al. Clinical and metabolic response to

    probiotic administration in patients with major depressive disorder: A randomized, double-blind,

    placebo-controlled trial. Nutrition 2016; 32(3):315-20.

    36- Whalen PJ, Rauch SL, Etcoff NL, et al. Masked presentations of emotional facial expressions

    modulate amygdala activity without explicit knowledge. J Neurosci 1998; 18(1): 411-8.

    37- Davis M. The role of the amygdala in fear and anxiety. Annu Rev Neurosci 1992;15: 353–75.

    38- Myers B, Greenwood-Van Meerveld B. Corticosteroid receptor-mediated mechanisms in the

    amygdala regulate anxiety and colonic sensitivity. Am J Physiol Gastrointest Liver Physiol 2007;

    292 (6): G1622-9.

    39- Bashashati M, Rezaei N, Shafieyoun A, et al. Cytokine imbalance in irritable bowel syndrome:

    a systematic review and meta-analysis. Neurogastroenterol Motil 2014; 26(7): 1036-48

    40- Haroon E, Raison CL, Miller AH. Psychoneuroimmunology meets neuropsycho-pharmacology:

    translational implications of the impact of inflammation on behavior. Neuropsychopharmacology

    2012; 37(1): 137-62.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    29

    41- Murray PS, Holmes PV. An overview of brain-derived neurotrophic factor and implications for

    excitotoxic vulnerability in the hippocampus. Int J Pept 2011; 2011: 654085.

    42- Duclot F, Kabbaj M. Epigenetic mechanisms underlying the role of brain-derived neurotrophic

    factor in depression and response to antidepressants. J Exp Biol 2015; 218(Pt 1): 21-31.

    43- Boulange CL, Neves AL, Chilloux J, et al. Impact of the gut microbiota on inflammation,

    obesity, and metabolic disease. Genome Med 2016;8:42.

    44- Selmer T, Andrei PI. p-Hydroxyphenylacetate decarboxylase from Clostridium difficile. A

    novel glycyl radical enzyme catalysing the formation of p-cresol. Eur J Biochem 2001; 268:1363-

    1372.

    45- Goodhart PJ, DeWolf WE Jr, Kruse LI. Mechanism-based inactivation of dopamine beta-

    hydroxylase by p-cresol and related alkylphenols. Biochemistry 1987;26:2576-83.

    46- Togsverd M, Werge TM, Tanko LB, et al. Association of a dopamine beta-hydroxylase gene

    variant with depression in elderly women possibly reflecting noradrenergic dysfunction. J Affect

    Disord 2008; 106:169-72.

    47- Kapoor A, Shandilya M, Kundu S. Structural insight of dopamine beta-hydroxylase, a drug

    target for complex traits, and functional significance of exonic single nucleotide polymorphisms.

    PLoS One 2011; 6:e26509.

    48- Johnston M, Pollard B, Hennessey P. Construct validation of the hospital anxiety and

    depression scale with clinical populations. J Psychosom Res 2000; 48(6): 579-84.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    30

    LEGENDS

    Figure 1: Flowchart study population

    Figure 2: Brain activation patterns assessed by fMRI

    Functional Magnetic Resonance Imaging (fMRI) was used to assess the BOLD response to fearful

    stimuli compared with fixation. Group differences in activation pattern are displayed. At baseline,

    when examining all brain regions (A), there was slightly greater engagement of the visual

    association (A1) and parietal cortices (A2) in B longum (BL) group compared with the placebo

    group, with no difference in predetermined region of interest (ROI), amygdala (B). After treatment

    (at 6 weeks), the BL group displayed lesser engagement of the amygdala, as well as frontal and

    temporal cortices that are involved in anxiety and mood regulation (C, D; in yellow-orange) and

    increased engagement of occipital regions (in blue) compared with the placebo group. At 6 week,

    the amygdala activation correlated with the depression scores in the whole cohort and the BL group

    (E, middle and right panel). Patients with adequate relief of IBS symptoms (blue dots) were more

    likely to have lesser engagement of the amygdala than the patients without improvement in their

    IBS symptoms (red dots).

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    Table 1: Demographics characteristics of study population

    B. longum n=22

    Placebo n=22

    Study status, n (%) Completed Dropped out

    18 (82) 4 (18)

    20 (91) 2 (9)

    Age, median (IQR) 46.5 (30-58) 40.0 (26-57) Female, n (%) 12 (54) 12 (54) Ethnicity n (%) Caucasian Other

    19 (86) 3 (14)

    21 (95) 1 (5)

    Smoking status Smokers, n ( %)

    3 (14) 3 (14)

    Alcohol consumption Consumers, n (%)

    8 (36) 11 (50)

    Fibre consumption, g/day, median (IQR)

    18.0 (12.0-23.2) 13.5 (10.0-18.2)

    BMI, median (IQR) 25.1 (21.5-28.4) 24.6 (22.3-29.5)

    IBS subtype n (%) Female Diarrhoea Female Mixed Male Diarrhea Male Mixed

    6 (27) 6 (27) 8 (37) 2 (9)

    6 (27) 6 (27) 7 (32) 3 (14)

    Anxiety and depression, n (%) Anxiety (HAD-A ≥8) Depression (HAD-D ≥8) Anxiety and depression

    21 (95) 13 (59) 12 (54)

    18 (82) 8 (36) 6 (27)

    No significant differences between groups for demographics characteristics (P value for all

    comparisons between groups >0.05)

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    Table 2. Depression, anxiety, IBS symptoms, quality of life and somatization scores

    Test/ Mean (SD) Before treatment Post-treatment, 6 weeks Mean difference

    95% CI Follow-up, 10 weeks Mean difference

    95% CI Placebo B. longum Placebo B. longum Placebo B. longum

    Depression HAD-D 5.2

    (3.0)

    7.6

    (3.7)

    4.5

    (3.1)

    3.9

    (3.1)

    0.6

    (-1.6 to 2.6)

    4.7

    (3.5)

    4.7

    (3.8)

    0.15

    (-2.5 to 2.5)

    Anxiety HAD-A 9.3 (2.6) 10.2

    (3.2)

    7.1

    (3.9)

    6.5

    (3.8)

    0.6

    (-1.9 to 3.2)

    8.0

    (4.3)

    7.6

    (4.8)

    0.39

    (-2.7 to 3.5)

    Anxiety STAI 40.4

    (12.3)

    41.3

    (13.9)

    38.8

    (12.9)

    33.1

    (9.5)

    5.7

    (-2.0 to 13.5)

    37.6

    (11.9)

    38.4

    (15.3)

    -0.83

    (-10.1 to 8.4)

    Anxiety TAI 44.0

    (11.9)

    47.7

    (10.5)

    42.5

    (11.3)

    39.4

    (11.6)

    3.1

    (-4.5 to 10.6)

    42.2

    (11.3)

    32.3

    (12.7)

    2.84

    (-5.2 to 10.9)

    IBS-Birmingham:

    total

    17.8

    (7.9)

    17.7

    (7.1)

    12.6

    (9.2)

    8.8

    (9.2)

    3.8

    (-2.4 to 9.9)

    13.0

    (6.9)

    12.4

    (9.5)

    0.55

    (-4.9 to 6.1)

    Birmingham:

    constipation

    3.8

    (3.6)

    3.0

    (3.1)

    3.1

    (3.5)

    1.4

    (1.6)

    1.7

    (-0.1 to 3.6)

    3.2

    (3.5)

    2.5

    (3.8)

    0.71

    (-1.7 to 3.2)

    Birmingham:

    diarrhea

    8.2

    (4.3)

    8.5

    (4.8)

    4.7

    (4.3)

    4.1

    (5.8)

    0.5

    (-2.8 to 3.9)

    5.1

    (3.2)

    5.2

    (4.9)

    -0.17

    (-2.9 to 2.6)

    Birmingham:

    Pain

    6.3

    (3.9)

    6.2

    (3.2)

    4.9

    (4.1)

    3.4

    (3.5)

    1.5

    (-1.1 to 4.1)

    4.7

    (3.4)

    4.7

    (3.8)

    0.15

    (-2.4 to 2.4)

    QoL-SF-36: Physical 43.9

    (10.8) 45.0

    (10.1) 43.1 (9.9)

    49.9 (8.8)

    -6.8 * (-13.2 to -0.4)

    46.9 (10.0)

    46.4 (9.6)

    0.52 (-6.3 to 7.4)

    QoL-SF-36 Mental 41.9

    (11.1)

    39.4

    (11.8)

    43.3

    (9.7)

    47.1

    (9.9)

    -3.80

    (-10.4 to 2.8)

    41.3

    (12.0)

    46.4

    (12.8)

    -5.06

    (-13.7 to 3.6)

    QoL-SF-36:

    Physical functioning

    78.5

    (22.5)

    78.3

    (24.2)

    76.8

    (22.7)

    94.5

    (9.5)

    -14.7*

    (-26.7 to -2.7)

    79.2

    (23.1)

    83.3

    (19.8)

    -4.17

    (-18.7 to 10.4)

    QoL-SF-36:

    Role physical

    51.3

    (38.4)

    61.1

    (43.1)

    47.5

    (38.8)

    80.8

    (35.9)

    -33.4*

    (-58.5 to -8.2)

    59.7

    (38.5)

    62.5

    (42.2)

    -2.77

    (-30.1 to 24.6)

    QoL-SF-36:

    Bodily pain

    61.2

    (24.2)

    51.0

    (20.1)

    58.2

    (23.3)

    65.1

    (20.7)

    -6.9

    (-21.7 to 7.9)

    62.2

    (25.5)

    60.1

    (20.4)

    2.11

    (-13.5 to 17.7)

    QoL-SF-36:

    General health

    55.2

    (19.2)

    59.6

    (20.9)

    60.9

    (22.1)

    68.1

    (17.0)

    -7.3

    (-20.6 to 6.1)

    59.9

    (20.7)

    65.6

    (21.1)

    -5.61

    (-19.8 to 8.5)

    QoL-SF-36: Vitality 45.5

    (19.2)

    40.3

    (18.7)

    43.7

    (18.9)

    55.3

    (17.4)

    -11.5

    (-23.8 to 0.7)

    46.4

    (20.4)

    49.7

    (25.0)

    -3.33

    (-18.8 to 12.1)

    QoL-SF-36:

    Social functioning

    61.9

    (21.6)

    56.9

    (26.8)

    68.1

    (24.4)

    77.2

    (19.4)

    -9.08

    (-24.0 to 5.8)

    63.9

    (26.7)

    72.2

    (22.5)

    -8.33

    (-25.1 to 8.4)

    QoL-SF-36:

    Role emotional

    60.0

    (39.9)

    55.6

    (39.6)

    56.7

    (40.6)

    78.4

    (38.9)

    -21.8

    (-48.5 to 4.9)

    59.3

    (43.6)

    68.5

    (43.5)

    -9.25

    (-38.7 to 20.2)

    QoL-SF-36:

    Mental health

    62.0

    (20.2)

    59.6

    (18.7)

    65.6

    (17.6)

    71.5

    (16.4)

    -5.9

    (-17.4 to 5.5)

    64.0

    (20.6)

    69.3

    (20.6)

    -5.33

    (-19.3 to 8.6)

    PHQ15: Somatization 11.1 11.9 10.0 8.4 1.61 9.4 10.0 -0.63

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    (2.9) (3.5) (3.9) (3.2) (-0.8 to 4.0) (3.5) (4.8) (-3.4 to 2.2)

    *p

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    Table 3: Decrease ≥2 points in depression and anxiety HAD scores at 6 and 10 weeks in the whole group (A) or when stratified by adequate relief of IBS symptoms (B)

    Outcome B. longum

    n Placebo

    n ITT Analysis

    RR 95% CI p value PP Analysis

    RR 95% CI p value

    Decrease HAD-D ≥2 at 6 weeks

    14 7 1.98 1.16-3.38 0.04 2.40 1.26-4.58 0.01

    Decrease HAD-A ≥2 at 6 weeks

    14 11 1.31 0.72-2.42 0.54 1.69 0.76-3.77 0.19

    Decrease HAD-D ≥2 at 10 weeks

    13 6 2.05 1.07-3.93 0.04 2.14 1.11-4.12 0.04

    Decrease HAD-A ≥2 at 10 weeks

    12 10 1.4 0.65-2.82 0.50 1.6 0.77-3.17 0.34

    Outcome

    B. longum n

    Placebo n

    ITT Analysis RR 95% CI p value

    PP Analysis RR 95% CI p value

    Adequate relief of IBS symptoms

    Decrease HAD-D ≥2 at 6 weeks

    11 2 3.07 0.89-10.6 0.03 3.07 0.89-10.6 0.03

    Decrease HAD-A ≥2 at 6 weeks

    11 4 1.53 0.72-3.27 0.66 1.53 0.72-3.27 0.34

    Decrease HAD-D ≥2 at 10 weeks

    4 1 6.00 1.03-35.9 0.04 6.00 1.06-35.9 0.04

    Decrease HAD-A ≥2 at 10 weeks

    4 4 1.5 0.85-2.64 0.46 1.50 0.85-2.64 0.46

    No Adequate relief of IBS symptoms

    Decrease HAD-D ≥2 at 6 weeks

    3 5 0.93 0.29-2.98 1.0 1.6 0.56-4.54 0.56

    Decrease HAD-A ≥2 at 6 weeks

    3 7 0.66 0.23-1.92 0.34 1.14 0.45-2.90 0.98

    Decrease HAD-D ≥2 at 10 weeks

    9 5 1.60 0.67-3.78 0.31 1.80 0.80-4.02 0.25

    A

    B-

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    Decrease HAD-A ≥2 at 10 weeks

    8 6 1.18 0.52-2.68 0.73 1.33 0.62-2.84 0.70

    Chi2 test, HAD-D: Hospital Anxiety and Depression score for depression, HAD-A: Hospital

    Anxiety and Depression score for anxiety, ITT: Intention-to-treat analysis, PP: Per-protocol

    analysis

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    Table 4: Brain activation patterns in all brain regions (top) and the amygdala (bottom).

    Left/right Brain region Brodmann

    area T-value p-value

    No.

    voxels

    Right Middle Temporal Gyrus BA 21 4.62796 0.000013 818

    Right Middle Temporal Gyrus BA 21 3.860474 0.000222 278

    Right Cerebellum * 3.964027 0.000154 452

    Right Inferior Frontal Gyrus BA 47 4.564617 0.000017 4657

    Right Cuneus BA 19 -3.889998 0.0002 1268

    Right Middle Occipital Gyrus BA 19 -4.455264 0.000026 898

    Right Middle Frontal Gyrus BA 10 4.099587 0.000095 526

    Right Amygdala/

    Parahippocampal Gyrus BA 28 3.983939 0.000144 1474

    Right Cuneus BA 19 -3.709217 0.000372 1527

    Right Medial Frontal Gyrus BA 11 4.73075 0.000009 365

    Left Middle Occipital Gyrus BA 19 -4.04685 0.000115 1196

    Left Amygdala/

    Parahippocampal Gyrus BA 35 4.451565 0.000026 1727

    Left Middle Frontal Gyrus BA 10 3.687655 0.0004 1080

    Left Middle Occipital Gyrus BA 19 -4.466287 0.000025 776

    Left Middle Temporal Gyrus BA 21 4.097827 0.000096 689

    Left Middle Occipital Gyrus BA 19 -3.282744 0.001499 472

    Left/right Brain region T-value p-value No. voxels

    Right Amygdala 3.98394 0.000144 690

    Left Amygdala 4.031566 0.000121 212

    Brain activation patterns assessed by fMRI for post-treatment group differences (placebo vs treatment; fear vs. fixation) in all brain regions (top) and for ROI- amygdala analyses (bottom). All data are corrected for multiple comparisons; FDR(q)

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    Table 5: Urinary metabolites after 6 weeks of treatment assessed by 1H NMR

    Urinary metabolites 1H NMR signal

    (multiplicity) OPLS Correlation

    coefficient (VIP)

    Relative concentration (au) p-value

    Placebo B. longum

    Creatine 3.04 (s) -0.44 (1.36) 14.49 (10.3) 7.44 (3.0) 0.007

    Phenylacetylglutamine 7.43 (m) -0.50 (1.76) 4.5 (1.8) 3.01 (1.3) 0.013

    4-cresol sulfate 2.35 (s) -0.36 (1.42) 5.38 (2.3) 3.9 (1.7) 0.022

    Trimethylamine-N-Oxide 3.27 (s) -0.32 (1.18) 44.0 (61.2) 15.48 (4.5) 0.002

    Metabolite data are reported as mean (SD) with an arbitrary unit (au) derived from 1H NMR

    spectral peak area. VIP: Variable Importance in Projection; m: multiplet; s: singlet.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    Figure 1.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    ROI - AmygdalaI

    -2 -1 0 1 2-10

    -5

    0

    5

    -2 -1 0 1 2-10

    -5

    0

    5

    -2 -1 0 1 2-10

    -5

    0

    5

    D amygdala activation (% BOLD)

    DH

    AD

    -D s

    core

    Pearson r=0.52, p=0.004

    D amygdala activation (% BOLD)D amygdala activation (% BOLD)

    DH

    AD

    -D s

    core

    DH

    AD

    -D s

    core

    Pooled Data BLPlacebo

    Pearson r=0.53, p=0.04

    Pearson r=0.14, p=0.02

    Adequate relief of IBS symptoms No adequate relief of IBS symptoms

    E

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    1

    Supplementary Figure 1: Design of the study

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    2

    Supplementary Figure 2: The backward masking paradigm.

    The backward masking paradigm: The fixation was jittered, followed by the presentation of a

    fearful or happy face for 67 ms, followed by the masking of the stimuli with a neutral face for 83

    ms. Participants had 1800 ms to respond. Four presentations of the paradigm were collected, with

    84 trials per presentation (scan).

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    3

    Supplementary Figure 3. Group responses to fear stimuli vs. happy or fixation stimuli.

    A. Full group response to fear face stimuli at both time points contrasted with the full group

    response to happy face stimuli. The analysis identified a number of brain regions that showed

    greater engagement in response to the fear stimuli including the amygdala, insula and regions in the

    frontal cortices.

    B. Full group response to fear stimuli contrasted with full group response to fixation stimuli, at both

    time points revealed broad activation including the fusiform gyri, thalamus, striatal regions, parietal

    cortices and hippocampal/amygdala complex.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    4

    Supplementary Figure 4: Gut microbiota analysis

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    5

    Supplementary Figure 5: Overview of 1H NMR urine metabolic profile and multivariate data

    analysis

    A: Typical 1H NMR urine metabolic profile with selected metabolite signature highlighted in

    aliphatic (0.5 – 4.5 ppm) and aromatic spectral areas (6.5 – 9.0 ppm).

    B: Overview of OPLS-DA coefficient plots resulting for urine NMR data and group discriminant

    analysis (placebo top, probiotics bottom). The OPLS coefficients plots are presented using a back-

    scaling transformation and projection to aid biomarker visualization. The direction of the signals in

    the plots relative to zero indicates positive or negative covariance with the group of interest. Each

    variable is plotted with a color code which indicates its discriminating power as calculated from the

    correlation matrix thus highlighting biomarker rich spectral regions.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    6

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    7

    Supplementary Figure 6: Differences in estimated marginal means for HAD-D and HAD-A

    (ANOVA multiple measurements)

    HAD-D: Hospital Anxiety and Depression score for depression, HAD-A: Hospital Anxiety and

    Depression score for anxiety

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    8

    Supplementary table 1: Full group responses at both time points in response to fear vs. happy stimuli

    Left/

    right Brain region Brodmann area T P value No. voxels

    Right Insula BA 13 5.4106 0.000008 3136

    Right Superior Temporal Gyrus BA 22 4.8403 0.00004 7845

    Right Inferior Temporal Gyrus BA 20 4.1807 0.000244 469

    Right Inferior Parietal Lobule BA 40 3.9513 0.000456 1144

    Right Superior Temporal Gyrus BA 21 3.3300 0.002375 159

    Right Middle Temporal Gyrus BA 22 3.7361 0.000815 1410

    Right Superior Temporal Gyrus BA 39 3.4202 0.001879 115

    Right Superior Temporal Gyrus BA 38 -4.6615 0.000065 181

    Right Cingulate Gyrus BA 24 4.4890 0.000105 1416

    Right Insula BA 13 3.3460 0.002279 186

    Right Lentiform Nucleus Lat. Globus

    Pallidus 4.7033 0.000058 4401

    Right Thalamus Pulvinar 4.6475 0.000067 1116

    Right Paracentral Lobule BA 5 4.2963 0.000178 855

    Left Parahippocampal Gyrus BA 27 4.3273 0.000163 306

    Left Precuneus BA 19 3.6435 0.001043 589

    Left Amygdala Amygdala 3.9502 0.000458 144

    Left Caudate Caudate Tail 3.3883 0.002041 156

    Left Insula BA 13 3.6715 0.000968 567

    Left Lentiform Nucleus Putamen 4.2270 0.000215 978

    Left Middle Occipital Gyrus BA 19 3.5165 0.00146 231

    Left Inferior Frontal Gyrus BA 46 3.4428 0.001771 142

    Left Middle Temporal Gyrus BA 21 3.78414 0.000717 505

    Left Superior Temporal Gyrus BA 22 4.2784 0.000187 683

    Left Inferior Temporal Gyrus BA 20 3.5100 0.001485 209

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    9

    Supplementary table 2: Inflammatory cytokines, BDNF and neurotransmitter levels at six weeks

    Test / Median (IQR)

    Baseline After treatment (6 weeks) U p

    B. longum Placebo B. longum Placebo

    CRP 1.12 (0.4-2.3) 0.85 (0.4-0.9) 1.32 (0.45-1.7) 1.70 (1.0-1.9) 120.5 0.18

    TNF-α 0.98 (0.82-1.17) 0.96 (0.89-1.25) 0.97 (0.82-1.05) 1.06 (0.95-1.29) 116.0 0.33

    IFN-γ 0.13 (0.07-0.22) 0.11 (0.08-0.15) 0.14 (0.10-0.18) 0.13 (0.09-0.18) 136.0 0.79

    IL-6 0.22 (0.14-0.27) 0.20 (0.15-0.24) 0.18 (0.13-0.26) 0.23 (0.18-0.40) 90.0 0.06

    IL-8 0.79 (0.64-1.42) 0.90 (0.65-1.17) 0.97 (0.70-1.14) 0.85 (0.66-1.11) 119.0 0.39

    IL-1β 0.97 (0.70-1.14) 0.05 (0.02-0.07) 0.04 (0.02-0.05) 0.04 (0.01-0.06) 116.0 0.33

    IL-10 1.32 (1.10-1.61) 1.39 (1.17-1.48) 1.45 (1.05-1.66) 1.69 (1.32-2.02) 101.0 0.14

    IL-12 p70 0.09 (0.04-0.15) 0.05 (0.04-0.07) 0.06 (0.05-0.08) 0.06 (0.04-0.09) 136.0 0.78

    IL10/12 ratio 26.5 (18.7-36.4) 24.1 (20.9-31.1) 19.7 (11.6-27.7) 24.4 (16.8-36.0) 114.0 0.30

    BDNF / β actin 20 (20-28) 29 (17-52) 25 (17-41) 37 (21-55) 124.0 0.10

    BDNF ELISA 6.61 (2.43-8.41) 4.87 (3.21-11.11) 7.00 (3.60-10.56) 5.83 (3.15-10.37) 148.0 0.65

    CGRP 23.6 (17.1-28.8) 25.2 (18.8-30.8) 23.8 (17.4-30.3) 26.5 (20.9-32.7) 147.0 0.34

    Substance P 1.01 (0.61-1.42) 1.26 (0.75-1.60) 1.03 (0.75-1.49) 1.19 (0.59-1.51) 176.0 0.91

    Serotonin 12.3 (5.8-14.5) 10.2 (6.6-19.3) 8.3 (4.2-14.9) 8.9 (5.9-12.1) 165.0 0.67

    Concentrations of individual biomarkers: CRP (mg/L), TNF-α, IFN-γ, IL-6, IL-8, IL-1β, IL-10 (all

    pg/mL), BDNF/β actin Log10-5 (copies/ng RNA), BDNF ELISA (ng/mL) CGRP (pg/mL), Substance P

    (ng/mL), Serotonin (ng/mL),

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    10

    Supplementary table 3: Most common adverse events

    Adverse Event B. longum n

    Placebo n

    P value Causality Outcome

    Constipation 0 2 0.48 NR NR

    Recovered Recovered

    Rectal bleeding 0 1 1.0 NR Recovered Rhinitis 2 1

    1.0 NR NR NR

    Recovered Recovered Recovered

    Headaches 4 1

    0.34

    Possible Possible Possible Possible NR

    Recovered Recovered Recovered Recovered Recovered

    Oral vesicles 1 0 1.0 NR Recovered Anal fissure 1 0 1.0 NR Recovered Neck pain 0 1 1.0 NR Recovered Urine infection 1 0 1.0 NR Dropped* Nausea 0 1 1.0 NR Recovered GERD symptoms 2 1 1.0 NR

    NR NR

    Recovered Recovered Recovered

    Abdominal pain 0 1 1.0 NR Recovered Diarrhoea 0 1 1.0 NR Recovered Cold 2 2 1.0 NR

    NR NR NR

    Recovered Recovered Recovered Recovered

    Otitis 1 0 1.0 NR Dropped* Food allergy 1 0 1.0 NR Recovered Streptococcus pharyngitis

    1 0 1.0 NR Dropped *

    Back pain 1 0 1.0 NR Recovered Iritis 1 0 1.0 NR Recovered Anxiety attack 0 1 1.0 NR Recovered

    Total 18 14 0.31

    NR: Not related, *Dropped from the study due to use of antibiotics

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    11

    Supplementary methods:

    Microbiota analysis:

    Microbiota analysis was performed using Illumina sequencing of the V3 region of 16S rRNA gene

    as described previously1. The data were processed by an in-house bioinformatics pipeline that

    incorporates quality filtering, Cutadapt2, PandaSeq3, AbundantOTU4, mothur5 and QIIME6.

    Abundant OTU provide output of clustered sequences in operational taxonomic units (OTUs).

    Taxonomic assignments use the RDP classifier7 with the Greengenes training set8. Analysis

    includes alpha-diversity measures for each sample and estimates of total diversity using QIIME, as

    well as β-diversity measures (weighted and unweighted Unifrac, Bray-Curtis) and other statistical

    analysis using QIIME and the PhyloSeq9 package implemented in R.

    Metabonomics analysis:

    1H NMR metabolite profiling approach was applied to urine samples, since this biofluid contains

    useful time-averaged representations of the recent homeostatic metabolic history of the individual

    and also carry indirect information on the gut microbial metabolic activities via the excretion

    patterns of many polar microbial-mammalian co-metabolites10, 11 (Supplementary Figure 5A).

    Forty µL of urine were mixed with 20 µL of deuterated phosphate buffer solution 0.6 M KH2PO4,

    containing 1 mM of sodium 3-(trimethylsilyl)-[2,2,3,3-2H4]-1-propionate (TSP, chemical shift

    reference δH = 0.0 ppm). 60µL of the mixture were transferred into 1.7mm NMR tubes. 1H NMR

    spectra were acquired with a Bruker Avance II 600 MHz spectrometer equipped with a 1.7 mm

    probe at 300 K (Bruker Biospin, Rheinstetten, Germany), using a standard pulse sequence with

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    12

    water suppression, and processed using TOPSPIN (version 2.1, Bruker, Germany) software

    package. The metabolite identification was achieved using in house database and 2D 1H NMR

    spectroscopy experiments.

    Chemometric analysis was performed using the software package SIMCA-P+ (version 14.0,

    Umetrics AB, Umeå, Sweden) and in-house developed MATLAB routines. Orthogonal Projection

    to Latent Structures (OPLS)12 and OPLS discriminant analysis (OPLS-DA) were employed for

    exploring the variance in the metabonomics data that may explain statistical differences between

    groups of samples. The classification accuracy of the OPLS-DA was established from the predicted

    samples in the 7-fold cross-validation cycle. To highlight the weight of individual variables in the

    model, Variable Importance in Projection (VIP) was used, with a value above 1 used as a threshold

    by convention. In addition, influential NMR variables that are correlated to the group separation are

    identified using the variable coefficients according to a previously published methods13.

    Representative signals of the identified metabolites were integrated and tested using non-parametric

    Mann Whitney test.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    13

    Additional references:

    1- Bartram AK, Lynch MD, Stearns JC, et al. Generation of multimillion-sequence 16S rRNA gene

    libraries from complex microbial communities by assembling paired-end illumina reads. Appl

    Environ Microbiol 2011, 77(11): 3846-3852.

    2- Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMB

    netjournal 2011, 17(1): 10-12.

    3- Masella AP, Bartram AK, Truszkowski JM, et al. PANDAseq: paired-end assembler for illumina

    sequences. BMC Bioinformatics 2012, 13: 31.

    4- Ye Y. Fast and accurate identification and quantification of abundant species from pyrosequences

    of 16S rRNA by consensus alignment. The proceedings of BIBM 2010: 153-157.

    5- Schloss PD, Westcott SL, Ryabin T, et al. Introducing mothur: open-source,platform-

    independent, community-supported software for describing and comparing microbial

    communities. Appl.Environ.Microbiol 2009, 75: 7537–7541.

    6- Caporaso JG, Kuczynski J, Stombaugh J, et al. QIIME allows analysis of high-throughput

    community sequencing data. Nat Methods 2010, 7(5): 335-336.

    7- Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of

    rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol 2007, 73:5261–5267.

    8- DeSantis TZ, Hugenholtz P, Larsen N, et al. Greengenes, a chimerachecked 16S rRNA gene

    database and workbench compatible with ARB. Appl Environ Microbiol 2006, 72:5069–5072.

  • MA

    NU

    SC

    RIP

    T

    AC

    CE

    PTE

    D

    ACCEPTED MANUSCRIPT

    14

    9- McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and

    graphics of microbiome census data. PloS one 2013, 8(4): e61217.

    10- Moco S, Collino S, Rezzi S, et al. Metabolomics perspectives in pediatric research. Pediatr Res

    2013;73:570-6.

    11- Collino S, Martin FP, Rezzi S. Clinical metabolomics paves the way towards future healthcare

    strategies. Br J Clin Pharmacol 2013;75:619-29.

    12- Trygg J, Wold S. O2-PLS, a two-block (X-Y) latent variable regression (LVR) method with an

    integrated OSC filter. J Chemom 2003;17:53-64.

    13- Cloarec O, Dumas ME, Craig A, et al. Statistical total correlation spectroscopy: an exploratory

    approach for latent biomarker identification from metabolic 1 H NMR data sets. Anal Chem

    2005;77:1282-1289.